Baricitinib: A Review of Pharmacology, Safety, and Emerging Clinical Experience in COVID-19

Sarah C J Jorgensen, Christopher L Y Tse, Lisa Burry, Linda D Dresser, Sarah C J Jorgensen, Christopher L Y Tse, Lisa Burry, Linda D Dresser

Abstract

A hyperinflammatory response to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection, reminiscent of cytokine release syndrome, has been implicated in the pathophysiology of acute respiratory distress syndrome and organ damage in patients with coronavirus disease 2019 (COVID-19). Agents that inhibit components of the pro-inflammatory cascade have garnered interest as potential treatment options with hopes that dampening the proinflammatory process may improve clinical outcomes. Baricitinib is a reversible Janus-associated kinase (JAK)-inhibitor that interrupts the signaling of multiple cytokines implicated in COVID-19 immunopathology. It may also have antiviral effects by targeting host factors that viruses rely for cell entry and by suppressing type I interferon driven angiotensin-converting-enzyme-2 upregulation. However, baricitinib's immunosuppressive effects may be detrimental during acute viral infections by delaying viral clearance and increasing vulnerability to secondary opportunistic infections. The lack of reliable biomarkers to monitor patients' immune status as illness evolves complicates deployment of immunosuppressive drugs like baricitinib. Furthermore, baricitinib carries the risk of increased thromboembolic events, which is concerning given the proclivity towards a hypercoagulable state in patients with COVID-19. In this article, we review available data on baricitinib with an emphasis on immunosuppressive and antiviral pharmacology, pharmacokinetics, safety, and current progress in COVID-19 clinical trials.

Keywords: COVID-19; JAK-inhibitor; Janus-associated kinase inhibitor; SARS-CoV-2; baricitinib; severe acute respiratory syndrome.

© 2020 Pharmacotherapy Publications, Inc.

Figures

Figure 1
Figure 1
Janus‐associated kinase/signal transducers and activators of transcription (JAK/STAT) pathway showing baricitinib inhibition of JAK complexes. GM‐CSF = granulocyte‐macrophage colony‐stimulating factor; IFNγ = interferon gamma. [Color figure can be viewed at wileyonlinelibrary.com]
Figure 2
Figure 2
Baricitinib inhibition of viral endocytosis. AAK1 = AP2‐associated protein kinase 1; ACE2 = angiotensin‐converting enzyme 2; GAK = G‐associated kinase; SARS‐CoV‐2 = severe acute respiratory syndrome‐coronavirus 2. [Color figure can be viewed at wileyonlinelibrary.com]

References

    1. Grasselli G, Zangrillo A, Zanella A, et al. Baseline characteristics and outcomes of 1591 patients infected with SARS‐CoV‐2 admitted to ICUs of the Lombardy Region, Italy. JAMA 2020;323:1574.
    1. Fu L, Wang B, Yuan T, et al. Clinical characteristics of coronavirus disease 2019 (COVID‐19) in China: a systematic review and meta‐analysis. J Infect 2020;80:656–665.
    1. Docherty AB, Harrison EM, Green CA, et al. Features of 20 133 UK patients in hospital with covid‐19 using the ISARIC WHO Clinical Characterisation Protocol: prospective observational cohort study. BMJ 2020;369:m1985.
    1. Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID‐19 in Wuhan, China: a retrospective cohort study. Lancet 2020;395(10229):1054–62.
    1. Mehta P, McAuley DF, Brown M, et al. COVID‐19: consider cytokine storm syndromes and immunosuppression. Lancet 2020;395(10229):1033–34.
    1. Peiris JS, Chu CM, Cheng VC, et al. Clinical progression and viral load in a community outbreak of coronavirus‐associated SARS pneumonia: a prospective study. Lancet 2003;361(9371):1767–72.
    1. Chen G, Wu D, Guo W, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020;130(5):2620–29.
    1. McGonagle D, Sharif K, O'Regan A, Bridgewood C. The role of cytokines including interleukin‐6 in COVID‐19 induced pneumonia and macrophage activation syndrome‐like disease. Autoimmun Rev 2020;19(6):102537.
    1. Jamilloux Y, Henry T, Belot A, et al. Should we stimulate or suppress immune responses in COVID‐19? Cytokine and anti‐cytokine interventions. Autoimmun Rev 2020;19(7):102567.
    1. Center for Drug Evaluation and Research . US Food and Drug Administration. Medical review baricitinib. NDA 207‐924. Apr. 2017. Available from . Accessed May 12, 2020.
    1. Favalli EG, Ingegnoli F, De Lucia O, Cincinelli G, Cimaz R, Caporali R. COVID‐19 infection and rheumatoid arthritis: faraway, so close!. Autoimmun Rev 2020;5:102523.
    1. Richardson P, Griffin I, Tucker C, et al. Baricitinib as potential treatment for 2019‐nCoV acute respiratory disease. Lancet 2020;10223:e30–1.
    1. Stebbing J, Phelan A, Griffin I, et al. COVID‐19: combining antiviral and anti‐inflammatory treatments. Lancet Infect Dis 2020;4:400–2.
    1. Kumar N, Sharma S, Kumar R, et al. Host‐directed antiviral therapy. Clin Microbiol Rev 2020;33(3):e00168‐19
    1. Samji H, Cescon A, Hogg RS, et al. Closing the gap: increases in life expectancy among treated HIV‐positive individuals in the United States and Canada. PLoS One 2013;12:e81355.
    1. Burstow NJ, Mohamed Z, Gomaa AI, et al. Hepatitis C treatment: where are we now? Int J Gen Med 2017;10:39–52.
    1. Jin M, Tong Q. Rhabdomyolysis as potential late complication associated with COVID‐19. Emerg Infect Dis 2020;26(7):1618–20.
    1. Fragoulis GE, McInnes IB, Siebert S. JAK‐inhibitors. New players in the field of immune‐mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford) 2019;58(Supp_1):i43–54.
    1. Pu SY, Xiao F, Schor S, et al. Feasibility and biological rationale of repurposing sunitinib and erlotinib for dengue treatment. Antiviral Res 2018;155:67–75.
    1. Richardson PJ, Corbellino M, Stebbing J. Baricitinib for COVID‐19: a suitable treatment? – Authors' reply. Lancet Infect Dis. Online ahead of print. 2020;S1473‐3099(20)30270‐X. 10.1016/S1473-3099(20)30270-X.
    1. Ghoreschi K, Laurence A, O'Shea JJ. Janus kinases in immune cell signaling. Immunol Rev 2009;1:273–87.
    1. Liu KD, Gaffen SL, Goldsmith MA. JAK/STAT signaling by cytokine receptors. Curr Opin Immunol 1998;3:271–8.
    1. Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol 2017;5:320.
    1. Center for Drug Evaluation and Research . US Food and Drug Administration. Pharmacology review baricitinib. NDA 207‐924. Apr. 2017. . Accessed 12 May 2020.
    1. Committee for Medicinal Products for Human Use . European Medicines Agency. Assessment report Olumiant (baricitinib). EMAE/H/C/004085/0000. 15 Dec. 2016. Available from . Accessed May 12, 2020.
    1. Center for Drug Evaluation and Research . US Food and Drug Administration. Clinical pharmacology and biopharmaceutics review baricitinib. NDA 207‐924. Apr. 2017. . Accessed 12 May 2020.
    1. Spinelli FR, Conti F, Gadina M. HiJAKing SARS‐CoV‐2? The potential role of JAK inhibitors in the management of COVID‐19. Sci Immunol 2020;5(47):eabc5367.
    1. Inoue Y, Tanaka N, Tanaka Y, et al. Clathrin‐dependent entry of severe acute respiratory syndrome coronavirus into target cells expressing ACE2 with the cytoplasmic tail deleted. J Virol 2007;16:8722–9.
    1. Kaksonen M, Roux A. Mechanisms of clathrin‐mediated endocytosis. Nat Rev Mol Cell Biol 2018;5:313–26.
    1. Hoffmann M, Kleine‐Weber H, Schroeder S, et al. SARS‐CoV‐2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020;181(2):271–80.e8.
    1. Bekerman E, Neveu G, Shulla A, et al. Anticancer kinase inhibitors impair intracellular viral trafficking and exert broad‐spectrum antiviral effects. J Clin Invest 2017;4:1338–52.
    1. Wang H, Yang P, Liu K, et al. SARS coronavirus entry into host cells through a novel clathrin‐ and caveolae‐independent endocytic pathway. Cell Res 2008;2:290–301.
    1. Ziegler CGK, Allon SJ, Nyquist SK, et al. SARS‐CoV‐2 receptor ACE2 is an interferon‐stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 2020;181:1016–35.e19.
    1. Vaduganathan M, Vardeny O, Michel T, McMurray JJV, Pfeffer MA, Solomon SD. Renin‐angiotensin‐aldosterone system inhibitors in patients with Covid‐19. N Engl J Med 2020;17:1653–59.
    1. Shi JG, Chen X, Lee F, et al. The pharmacokinetics, pharmacodynamics, and safety of baricitinib, an oral JAK 1/2 inhibitor, in healthy volunteers. J Clin Pharmacol 2014;12:1354–61.
    1. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of Covid‐19 – preliminary report. N Engl J Med 2020;NEJMoa2007764. Online ahead of print. 10.1056/NEJMoa2007764.
    1. Meißner Y, Baganz L, Schneider M. Baricitinib and tofacitinib in real life – does obesity impact response to janus kinase inhibitor therapy in rheumatoid arthritis? [abstract]. Arthritis Rheumatol 2018;70(suppl 10). . Accessed May 12, 2020.
    1. Kim H, Brooks KM, Tang CC, et al. Pharmacokinetics, pharmacodynamics, and proposed dosing of the oral JAK1 and JAK2 inhibitor baricitinib in pediatric and young adult CANDLE and SAVI patients. Clin Pharmacol Ther 2018;2:364–73.
    1. Arthritis Advisory Committee Meeting FDA Briefing Document. NDA 207924. Baricitinib Janus Kinase (JAK) inhibitor for RA. Eli Lilly and Company (Lilly). 23 April 2018. Available from . Accessed May 15, 2020.
    1. . Available from . Accessed May 18, 2020.
    1. National Institutes of Health . US Department of Health and Human Services. News release. NIH clinical trial testing antiviral remdesivir plus anti‐inflammatory drug baricitinib for COVID‐19 begins. 8 May 2020. . Accessed 26 May 2020.
    1. Cantini F, Niccoli L, Matarrese D, Nicastri E, Stobbione P, Goletti D. Baricitinib therapy in COVID‐19: a pilot study on safety and clinical impact. J Infect 2020;Online ahead of print. : S0163‐4453(20)30228‐0. 10.1016/j.jinf.2020.04.017.
    1. Vyas D, O'Dell KM, Bandy JL, Boyce EG. Tofacitinib: the First Janus Kinase (JAK) inhibitor for the treatment of rheumatoid arthritis. Ann Pharmacother 2013;11:1524–31.
    1. Qin C, Zhou L, Hu Z, et al. Dysregulation of immune response in patients with COVID‐19 in Wuhan, China. Clin Infect Dis 2020. Mar 12;ciaa248. Online ahead of print. 10.1093/cid/ciaa248.
    1. Wang D, Hu B, Hu C, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus‐infected pneumonia in Wuhan, China. JAMA 2020;323:1061.
    1. Zheng M, Gao Y, Wang G, et al. Functional exhaustion of antiviral lymphocytes in COVID‐19 patients. Cell Mol Immunol 2020;5:533–35.
    1. Guo T Fan Y, Chen M, et al. Cardiovascular implications of fatal outcomes of patients with coronavirus disease 2019 (COVID‐19). JAMA Cardiol 2020; e201017. Online ahead of print. 10.1001/jamacardio.2020.1017.
    1. Anjara P, Jiang M, Mundae M. Symptomatic elevation creatine kinase following treatment of rheumatoid arthritis with baricitinib. Clin Rheumatol 2020;2:613–14.
    1. Queeney K, Housley W, Sokolov J, Long A. Elucidating the mechanism underlying creatine phosphokinase upregulation with upadacitinib. Annual European Congress of Rheumatology, EULAR 2019, Madrid, 12–15 June 2019. FRIO131. 10.1136/annrheumdis-2019-eular.7509.
    1. Center for Drug Evaluation and Research . US Food and Drug Administration. Medical review tofacitinib. NDA 203‐214. Aug. 2012. . Accessed 15 May 2020.
    1. Center for Drug Evaluation and Research . US Food and Drug Administration. Clinical review upadacitinib. NDA 211‐675. Dec. 2019. . Accessed 15 May 2020.
    1. Grisouard J, Hao‐Shen H, Dirnhofer S, Wagner KU, Skoda RC. Selective deletion of Jak2 in adult mouse hematopoietic cells leads to lethal anemia and thrombocytopenia. Haematologica 2014;4:e52–4.
    1. Meyer SC, Keller MD, Woods BA, et al. Genetic studies reveal an unexpected negative regulatory role for Jak2 in thrombopoiesis. Blood 2014;14:2280–4.
    1. Ng AP, Kauppi M, Metcalf D, et al. Mpl expression on megakaryocytes and platelets is dispensable for thrombopoiesis but essential to prevent myeloproliferation. Proc Natl Acad Sci USA 2014;16:5884–9.
    1. Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost 2020;4:844–47.
    1. Yin S, Huang M, Li D, Tang N. Difference of coagulation features between severe pneumonia induced by SARS‐CoV2 and non‐SARS‐CoV2. J Thromb Thrombolysis 2020:1–4.
    1. Panigada M, Bottino N, Tagliabue P, et al. Hypercoagulability of COVID‐19 patients in intensive care unit. A report of thromboelastography findings and other parameters of hemostasis. J Thromb Haemost 2020;18(7):1738–42. Epub 2020 Jun 24. 10.1111/jth.14850.
    1. Hadjadj J, Yatim N, Barnabei L, et al. Impaired type I interferon activity and exacerbated inflammatory responses in severe Covid‐19 patients. medRxiv 2020;2020.04.19.20068015.
    1. Trouillet‐Assant S, Viel S, Gaymard A, et al. Type I IFN immunoprofiling in COVID‐19 patients. J Allergy Clin Immunol 2020;146(1):206–208.e2. 10.1016/j.jaci.2020.04.029.
    1. Rawson TM, Moore LSP Zhu N, et al. Bacterial and fungal co‐infection in individuals with coronavirus: A rapid review to support COVID‐19 antimicrobial prescribing. Clin Infect Dis 2020; ciaa530.. Online ahead of print. 10.1093/cid/ciaa530.
    1. Sepulveda J, Westblade LF, Whittier S, et al. Bacteremia and blood culture utilization during COVID‐19 surge in New York City. J Clin Microbiol 2020. May 13;JCM.00875‐20. Online ahead of print. 10.1128/JCM.00875-20.
    1. Vallejo‐Yague E, Weiler S, Micheroli R, Burden AM. Thromboembolic safety reporting of tofacitinib and baricitinib: An analysis of the WHO VigiBase. Online ahead of print. Drug Saf. 2020. Jun 12; Online ahead of print. 10.1007/s40264-020-00958-9.
    1. Product monograph Olumiant (baricitinib) tablets . Eli Lilly Canada Inc. April 2020. . Accessed 15 May 2020.
    1. Connors JM, Levy JH. Thromboinflammation and the hypercoagulability of COVID‐19. J Thromb Haemost 2020;18(7):1559–1561.
    1. Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic treatments for coronavirus disease 2019 (COVID‐19): a review. JAMA 2020. Apr 13. Online ahead of print. 10.1001/jama.2020.6019.
    1. Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis 2013;3:260–8.
    1. Kox WJ, Volk T, Kox SN, Volk HD. Immunomodulatory therapies in sepsis. Intensive Care Med 2000;S124–8.
    1. Ranieri VM, Thompson BT, Barie PS, et al. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med 2012;22:2055–64.
    1. Abraham E, Anzueto A, Gutierrez G, et al. Double‐blind randomised controlled trial of monoclonal antibody to human tumour necrosis factor in treatment of septic shock. Lancet 1998;351(9107):929–33.
    1. Cronin L, Cook DJ, Carlet J, et al. Corticosteroid treatment for sepsis: a critical appraisal and meta‐analysis of the literature. Crit Care Med 1995;8:1430–9.
    1. Dhainaut JF, Vincent JL, Richard C, et al. CDP571, a humanized antibody to human tumor necrosis factor‐alpha: safety, pharmacokinetics, immune response, and influence of the antibody on cytokine concentrations in patients with septic shock. CPD571 Sepsis Study Group. Crit Care Med 1995;9:1461–9.

Source: PubMed

3
订阅